88 research outputs found

    Oncostatin M Promotes Breast Cancer Metastasis: Increased Expression of Pro-Angiogenic Factors, Inflammatory Cytokine Expression, and Circulating Tumor Cell Numbers

    Get PDF
    Breast cancer is the most diagnosed cancer type in women and its resultant mortality is second only to lung cancer worldwide. While breast cancer is known to have many risk factors, inflammation remains an unquantifiable risk, and it can arise from obesity, depression, poor health, autoimmune diseases, and other conditions that cause systemic chronic inflammation. Chronic inflammation is gaining recognition for its role in cancer development, the potentiation of a metastatic phenotype in cancer cells, and decreased survival in breast cancer patients. In particular, inflammatory cytokines in the interleukin-6 (IL-6) family have been shown to promote an epithelial-mesenchymal transition (EMT), tumor cell detachment, invasion, and metastasis. However, therapies to inhibit IL-6 have not been successful in treating solid tumors. This is most likely due to redundancy, as there are other inflammatory cytokines such as oncostatin M (OSM) and interleukin-1 beta (IL-1β) that demonstrate overlapping effects in cancer progression. In these studies, the interactions between OSM, IL-6 and IL-1β were addressed. First, OSM and IL-6 were shown to induce vascular endothelial growth factor (VEGF) in a breast cancer subtype-specific manner. Next, OSM was assessed for its capacity to increase circulating tumor cell numbers in mouse models of human breast cancer. Lastly, OSM, IL-6, and IL-1β expression levels were shown to correlate with each other in breast cancer, and high co-expression of these cytokines was shown to lead to decreased patient survival. Furthermore, OSM was assessed for its synergistic relationship with IL-1β in inducing IL-6 secretion from breast cancer cells. Together, these results suggest that inflammatory cytokines promote metastatic disease in a breast cancer subtype-dependent manner. Importantly, these studies both provide a rationale for the development of breast cancer therapeutic regimens that target multiple cytokines as well as help explain why single anti-cytokine therapies have failed in clinical trials

    Progression of Breast Cancer Metastatic Disease and Subsequent Osteolytic Bone Degradation Mediated by Oncostatin M

    Get PDF
    Breast cancer is the most diagnosed cancer in women and is the second most common cancer-related death for women worldwide. While the primary tumor itself is not lethal, the metastases that disrupt vital organ functions pose a significant clinical challenge. Seventy percent of women with metastatic breast cancer have metastases to the bone, which is the most significant cause of morbidity for these patients. Oncostatin M (OSM) is a pleiotropic cytokine that plays a role in the immune system, wound repair, and haematopoiesis. OSM was previously considered for anticancer therapy due to its anti-proliferative effects against breast cancer cells in vitro. However, recent studies in the literature and from our lab suggest that OSM increases the metastatic potential of breast cancer cells. OSM has been shown to increase angiogenesis through the induction of VEGF and invasion through the release of MMP family of proteases. However, the exact role that OSM has on the metastatic cascade of breast cancer remains unclear. In this study, we attempted to elucidate the role of OSM on breast cancer metastases in an in vivo and in an in vitro mouse model of breast cancer. The results indicate that OSM increases pro-metastatic characteristics on 4T1.2 murine mammary cancer cells in vitro. OSM induced detachment and various factors that are thought to promote metastases and bone degredation such as VEGF, COX-2, IL-6, and HIF1α;. In an in vivo orthotopic 4T1.2 mouse model of breast cancer, OSM also increased the metastatic burden to the lung, spleen and the liver in vivo, while tumor growth was unaffected. In an in vitro co-culture model of the metastatic bone microenvironment, OSM and murine mammary cancer cells synergistically increased osteoclast differentiation and activity. Furthermore, inhibition of VEGF, COX-2, IL-6, or HIF1α; attenuates osteoclast differentiation. Our data suggest that OSM might be a useful target for individualized anticancer therapies on cancer patients with high level of serum OSM concentrations and may help prevent metastases and bone destruction in breast

    Clinical significance of interleukin (IL)-6 in cancer metastasis to bone: potential of anti-IL-6 therapies

    Get PDF
    Metastatic events to the bone occur frequently in numerous cancer types such as breast, prostate, lung, and renal carcinomas, melanoma, neuroblastoma, and multiple myeloma. Accumulating evidence suggests that the inflammatory cytokine interleukin (IL)-6 is frequently upregulated and is implicated in the ability of cancer cells to metastasize to bone. IL-6 is able to activate various cell signaling cascades that include the STAT (signal transducer and activator of transcription) pathway, the PI3K (phosphatidylinositol-3 kinase) pathway, and the MAPK (mitogen-activated protein kinase) pathway. Activation of these pathways may explain the ability of IL-6 to mediate various aspects of normal and pathogenic bone remodeling, inflammation, cell survival, proliferation, and pro-tumorigenic effects. This review article will discuss the role of IL-6: 1) in bone metabolism, 2) in cancer metastasis to bone, 3) in cancer prognosis, and 4) as potential therapies for metastatic bone cancer

    Novel Mouse Mammary Cell Lines for \u3cem\u3ein vivo\u3c/em\u3e Bioluminescence Imaging (BLI) of Bone Metastasis

    Get PDF
    Background Tumor cell lines that can be tracked in vivo during tumorigenesis and metastasis provide vital tools for studying the specific cellular mechanisms that mediate these processes as well as investigating therapeutic targets to inhibit them. The goal of this study was to engineer imageable mouse mammary tumor cell lines with discrete propensities to metastasize to bone in vivo. Two novel luciferase expressing cell lines were developed and characterized for use in the study of breast cancer metastasis to bone in a syngeneic mouse model. Results The 4T1.2 luc3 and 66c14 luc2 cell lines were shown to have high levels of bioluminescence intensity in vitro and in vivo after orthotopic injection into mouse mammary fat pads. The 4T1.2 luc3 cell line was found to closely model the sites of metastases seen in human patients including lung, liver, and bone. Specifically, 4T1.2 luc3 cells demonstrated a high incidence of metastasis to spine, with an ex-vivo BLI intensity three orders of magnitude above the commercially available 4T1 luc2 cells. 66c14 luc2 cells also demonstrated metastasis to spine, which was lower than that of 4T1.2 luc3 cells but higher than 4T1 luc2 cells, in addition to previously unreported metastases in the liver. High osteolytic activity of the 4T1.2 luc3 cells in vivo in the bone microenvironment was also detected. Conclusions The engineered 4T1.2 luc3 and 66c14 luc2 cell lines described in this study are valuable tools for studying the cellular events moderating the metastasis of breast tumor cells to bone

    Oncostatin M Promotes Mammary Tumor Metastasis to Bone and Osteolytic Bone Degradation

    Get PDF
    Oncostatin M (OSM) is an interleukin-6 (IL-6) family cytokine that has been implicated in a number of biological processes including inflammation, hematopoiesis, immune responses, development, and bone homeostasis. Recent evidence suggests that OSM may promote breast tumor invasion and metastasis. We investigated the role of OSM in the formation of bone metastases in vivo using the 4T1.2 mouse mammary tumor model in which OSM expression was knocked down using shRNA (4T1.2-OSM). 4T1.2-OSM cells were injected orthotopically into Balb/c mice, resulting in a greater than 97% decrease in spontaneous metastasis to bone compared to control cells. Intratibial injection of these same 4T1.2-OSM cells also dramatically reduced the osteolytic destruction of trabecular bone volume compared to control cells. Furthermore, in a tumor resection model, mice bearing 4T1.2-OSM tumors showed an increase in survival by a median of 10 days. To investigate the specific cellular mechanisms important for OSM-induced osteolytic metastasis to bone, an in vitro model was developed using the RAW 264.7 preosteoclast cell line co-cultured with 4T1.2 mouse mammary tumor cells. Treatment of co-cultures with OSM resulted in a 3-fold induction of osteoclastogenesis using the TRAP assay. We identified several tumor cell–induced factors including vascular endothelial growth factor, IL-6, and a previously uncharacterized OSM-regulated bone metastasis factor, amphiregulin (AREG), which increased osteoclast differentiation by 4.5-fold. In addition, pretreatment of co-cultures with an anti-AREG neutralizing antibody completely reversed OSM-induced osteoclastogenesis. Our results suggest that one mechanism for OSM-induced osteoclast differentiation is via an AREG autocrine loop, resulting in decreased osteoprotegerin secretion by the 4T1.2 cells. These data provide evidence that OSM might be an important therapeutic target for the prevention of breast cancer metastasis to bone

    Novel mouse mammary cell lines for in vivo bioluminescence imaging (BLI) of bone metastasis

    Get PDF
    Abstract Background Tumor cell lines that can be tracked in vivo during tumorigenesis and metastasis provide vital tools for studying the specific cellular mechanisms that mediate these processes as well as investigating therapeutic targets to inhibit them. The goal of this study was to engineer imageable mouse mammary tumor cell lines with discrete propensities to metastasize to bone in vivo. Two novel luciferase expressing cell lines were developed and characterized for use in the study of breast cancer metastasis to bone in a syngeneic mouse model. Results The 4 T1.2 luc3 and 66c14 luc2 cell lines were shown to have high levels of bioluminescence intensity in vitro and in vivo after orthotopic injection into mouse mammary fat pads. The 4 T1.2 luc3 cell line was found to closely model the sites of metastases seen in human patients including lung, liver, and bone. Specifically, 4 T1.2 luc3 cells demonstrated a high incidence of metastasis to spine, with an ex-vivo BLI intensity three orders of magnitude above the commercially available 4 T1 luc2 cells. 66c14 luc2 cells also demonstrated metastasis to spine, which was lower than that of 4 T1.2 luc3 cells but higher than 4 T1 luc2 cells, in addition to previously unreported metastases in the liver. High osteolytic activity of the 4 T1.2 luc3 cells in vivo in the bone microenvironment was also detected. Conclusions The engineered 4 T1.2 luc3 and 66c14 luc2 cell lines described in this study are valuable tools for studying the cellular events moderating the metastasis of breast tumor cells to bone.</p

    Oncostatin M Binds to Extracellular Matrix in a Bioactive Conformation: Implications for Inflammation and Metastasis

    Get PDF
    Oncostatin M (OSM) is an interleukin-6-like inflammatory cytokine reported to play a role in a number of pathological processes including cancer. Full-length OSM is expressed as a 26 kDa protein that can be proteolytically processed into 24 kDa and 22 kDa forms via removal of C-terminal peptides. In this study, we examined both the ability of OSM to bind to the extracellular matrix (ECM) and the activity of immobilized OSM on human breast carcinoma cells. OSM was observed to bind to ECM proteins collagen types I and XI, laminin, and fibronectin in a pH-dependent fashion, suggesting a role for electrostatic bonds that involves charged amino acids of both the ECM and OSM. The C-terminal extensions of 24 kDa and 26 kDa OSM, which contains six and thirteen basic amino acids, respectively, enhanced electrostatic binding to ECM at pH 6.5–7.5 when compared to 22 kDa OSM. The highest levels of OSM binding to ECM, though, were observed at acidic pH 5.5, where all forms of OSM bound to ECM proteins to a similar extent. This indicates additional electrostatic binding properties independent of the OSM C-terminal extensions. The reducing agent dithiothreitol also inhibited the binding of OSM to ECM suggesting a role for disulfide bonds in OSM immobilization. OSM immobilized to ECM was protected from cleavage by tumor-associated proteases and maintained activity following incubation at acidic pH for extended periods of time. Importantly, immobilized OSM remained biologically active and was able to induce and sustain the phosphorylation of STAT3 in T47D and ZR-75-1 human breast cancer cells over prolonged periods, as well as increase levels of STAT1 and STAT3 protein expression. Immobilized OSM also induced epithelial–mesenchymal transition-associated morphological changes in T47D cells. Taken together, these data indicate that OSM binds to ECM in a bioactive state that may have important implications for the development of chronic inflammation and tumor metastasis

    OSM-induced CD44 contributes to breast cancer metastatic potential through cell detachment but not epithelial-mesenchymal transition

    Get PDF
    Background: Hormone receptor status in human breast cancer cells is a strong indicator of the aggressiveness of a tumor. Triple negative breast cancers (TNBC) are aggressive, difficult to treat, and contribute to high incidences of metastasis by possessing characteristics such as increased tumor cell migration and a large presence of the transmembrane protein, cluster of differentiation 44 (CD44) on the cell membrane. Estrogen receptor-positive (ER+) cells are less aggressive and do not migrate until undergoing an epithelial-mesenchymal transition (EMT). Methods: The relationship between EMT and CD44 during metastatic events is assessed by observing changes in EMT markers, tumor cell detachment, and migration following cytokine treatment on both parental and CD44 knockdown human breast tumor cells. Results: ER+ T47D and MCF-7 human breast cancer cells treated with OSM demonstrate increased CD44 expression and CD44 cleavage. Conversely, ER- MDA-MB-231 human breast cancer cells do not show a change in CD44 expression nor undergo EMT in the presence of OSM. In ER+ cells, knockdown expression of CD44 by shRNA did not prevent EMT but did change metastatic processes such as cellular detachment and migration. OSM-induced migration was decreased in both ER+ and ER- cells with shCD44 cells compared to control cells, while the promotion of tumor cell detachment by OSM was decreased in ER+ MCF7-shCD44 cells, as compared to control cells. Interestingly, OSM-induced detachment in ER- MDA-MB-231-shCD44 cells that normally don't detach at significant rates. Conclusion: OSM promotes both EMT and tumor cell detachment in ER+ breast cancer cells. Yet, CD44 knockdown did not affect OSM-induced EMT in these cells, while independently decreasing OSM-induced cell detachment. These results suggest that regulation of CD44 by OSM is important for at least part of the metastatic cascade in ER+ breast cancer.The following people have contributed to this work in more ways than one. Raquel Brown provided great insight and technical experience for immunofluorescent imaging. Hannah Scott provided data analysis and contributed to the growth and propagation of cells used for this study. This study was partially funded by the following grants: NIH/NCI R15CA137510, NIH/NCRR P20RR016454, NIH/NIGMS P20GM103408, NIH/NIGMS P20GM109095, Susan G. Komen Foundation KG100513, and American Cancer Society RSG-09-276-01-CSM.S

    Co-Expression of VEGF and IL-6 Family Cytokines is Associated with Decreased Survival in HER2 Negative Breast Cancer Patients: Subtype-Specific IL-6 Family Cytokine-Mediated VEGF Secretion

    Get PDF
    Breast cancer cell-response to inflammatory cytokines such as interleukin-6 (IL-6) and oncostatin M (OSM) may affect the course of clinical disease in a cancer subtype-dependent manner. Furthermore, vascular endothelial growth factor A (VEGF) secretion induced by IL-6 and OSM may also be subtype-dependent. Utilizing datasets from Oncomine, we show that poor survival of invasive ductal carcinoma (IDC) breast cancer patients is correlated with both high VEGF expression and high cytokine or cytokine receptor expression in tumors. Importantly, epidermal growth factor receptor-negative (HER2-), but not HER2-positive (HER2+), patient survival is significantly lower with high tumor co-expression of VEGF and OSM, OSMRβ, IL-6, or IL-6Rα compared to low co-expression. Furthermore, assessment of HER2- breast cancer cells in vitro identified unique signaling differences regulating cytokine-induced VEGF secretion. The levels of VEGF secretion were analyzed by ELISA with siRNAs for hypoxia inducible factor 1 α (HIF1α) and signal transducer and activator of transcription 3 (STAT3). Specifically, we found that estrogen receptor-negative (ER-) MDA-MB-231 cells respond only to OSM through STAT3 signaling, while ER+ T47D cells respond to both OSM and IL-6, though to IL-6 to a lesser extent. Additionally, in the ER+ T47D cells, OSM signals through both STAT3 and HIF1α. These results highlight that the survival of breast cancer patients with high co-expression of VEGF and IL-6 family cytokines is dependent on breast cancer subtype. Thus, the heterogeneity of human breast cancer in relation to IL-6 family cytokines and VEGF may have important implications in clinical treatment options, disease progression, and ultimately patient prognosis

    Comparison of energy metabolism in Insulin-Dependent and Non-Insulin-Dependent diabetes mellitus

    Get PDF
    To compare the metabolic consequences of insulin-dependent diabetes mellitus (IDDM) and non-insulin-dependent diabetes mellitus (NIDDM), glycemic control and energy metabolism were evaluated in 18 children displaying IDDM and 19 NIDDM adult patients. With rising concentrations of fasting blood glucose (FBG), hemoglobin A1C and free fatty acid, the percentage of the ratio of resting energy expenditure (REE) to predicted REE expressed as %REE increased and the respiratory quotient (RQ) decreased. The linear regression between RQ and FBG showed the same gradient in IDDM and NIDDM although the RQ in IDDM was always 0.07 lower than that in NIDDM given various FBG concentrations. Those patients whose RQ values were less than 0.7, indicating ketone body production, included 8 (44%) IDDM and 2 (11%) NIDDM patients. These results may explain the relatively greater manifestation of ketoacidosis in IDDM
    corecore